All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know AML.

The AML Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your AML Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The AML Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the AML Hub cannot guarantee the accuracy of translated content. The AML Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2020-04-09T14:31:10.000Z

Editorial theme | Use of checkpoint inhibitors in AML therapy

Bookmark this article

Acute myeloid leukemia (AML) is a hematological cancer characterized by uncontrolled growth of abnormal myeloblasts that infiltrate the bone marrow, blood, and other tissues.In 1973, for the first time, the induction regimen known as “7+3” (anthracycline + cytarabine) was reported, and for over four decades it has been the standard induction regimen for AML. However, since 2017, there has been an explosion of new treatment options, in particular therapeutic agents targeting specific subtypes of AML.2

Given the important role of the immune system in cancer eradication, immunotherapies such as vaccines, drug-conjugated and bispecific monoclonal antibodies, cell-based therapy, and immune checkpoint inhibitors are being evaluated.3 Here, we provide an overview of the use of checkpoint inhibitors in AML therapy — the next AML Hub editorial theme.

The use of immune checkpoint inhibitors, such as anti-PD-1, anti-PD-L1, or anti-CTLA-4, in combination with chemotherapy or hypomethylating agents (HMAs) is promising.4 At the 2018 American Society of Hematology Annual Meeting, Jacalyn Rosenblatt discussed the scientific rationale for incorporating checkpoint inhibitors in AML therapy, suggesting the following reasons for the use of checkpoint inhibitors at remission (see full article here):

  • The observed increase of PD-L1 expression in the bone marrow (BM) of patients with TP53 mutations and patients with adverse cytogenetics, compared to healthy controls
  • The apparent increase of TIM-3 (immune checkpoint molecule) expression on CD4 and CD8 T cells in the BM and high PD-1 in peripheral CD4+ T cells in patients achieving remission

At the 44th European Society for Blood and Marrow Transplantation (EBMT) Annual Meeting, Arnon Nagler discussed the use of checkpoint inhibitors with the AML Hub (video below). His focus was nivolumab, a PD-1 inhibitor, for the prevention of posttransplantation relapse in patients with AML.

VIDEO INTERVIEW: Prof. Arnon Nagler | EBMT 2018 | Prospects of immune checkpoint inhibitors for prevention of SCT relapse in AML

Prospects of immune checkpoint inhibitors for prevention of SCT relapse in AML

There are several completed and ongoing clinical trials evaluating the benefit of immune checkpoint inhibitors, alone and in combination, in patients with AML.4 Results of clinical trials on immune checkpoint inhibitors as monotherapy are reported in Table 1.

Table 1. Clinical trials on immune checkpoint inhibitors as monotherapy

ALL, acute lymphoblastic leukemia; allo-SCT, allogeneic stem cell transplantation; AML, acute myeloid leukemia; CR, complete response; MDS, myelodysplastic syndrome; MoA, mechanism of action

Investigational medication

MoA

N

Phase

Indication

Results

CT-011

Anti-PD-1 antibody

17
(8 with AML)

I

Hematologic cancer

Only one of the eight AML patients responded after the first dose of CT-011, with a reduction in peripheral blasts (50% to 5%)

Ipilimumab

Anti-CTLA-4 antibody

28
(12 with AML)

I

NCT01822509

Hematologic malignancies relapsed after allo-SCT

CR observed in five patients (four with extramedullary AML and one with MDS developing into AML) treated with 10 mg per kilogram of body weight

Ongoing clinical trials

Pembrolizumab

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Anti-PD-1 antibody

 

I

NCT03291353

Refractory AML

 

I

NCT02981914

Hematologic malignancies (including AML) relapsed after allo-SCT

I

NCT03286114

AML/MDS/ALL relapsed after allo-SCT

II

NCT02708641

AML post remission in patients ≥ 60 years transplant ineligible

Pembrolizumab + autologous SCT

Anti-PD-1 antibody

II

NCT02771197

AML with high risk of relapse ineligible for allo-SCT

Nivolumab

Anti-PD-1 antibody

II

NCT02275533

AML in CR

II

NCT02532231

AML in CR at high risk of relapse

Chemotherapeutic agents, such as anthracyclines, are inducers of immunogenic cell death, which leads to cytotoxic T lymphocyte (CTLs) proliferation and interferon-gamma (INF-γ) release. INF-γ release leads to PD-L1 increased expression on leukemic blasts. The increased expression of PD-L1 limits the ability of CTLs to eradicate leukemic blasts, resulting in resistant leukemia cells that may cause a relapse of the disease. The combination immune checkpoint inhibitors/chemotherapy could represent a good strategy to overcome this resistance and several clinical trials are testing it. Clinical trials on immune checkpoint inhibitors plus chemotherapy are reported in Table 2.

Table 2. Clinical trials on immune checkpoint inhibitors in combination with chemotherapy

AML, acute myeloid leukemia; CR, complete response; CRi, complete response with incomplete count recovery; MDS, myelodysplastic syndrome; R/R, relapsed/refractory 

Investigational regimen

N

Phase

Indication

Results

Nivolumab + cytarabine/ idarubicin

32
(30 with AML)

II

Upfront therapy AML, high-risk MDS

 

CR/CRi 72%

High-dose cytarabine + pembrolizumab followed by pembrolizumab maintenance therapy in case of response

13

II

R/R AML

CR/CRi 40%

Ongoing clinical trials

Nivolumab + cyclophosphamide

 

I/II

NCT03417154

R/R AML, high-risk MDS

 

Pembrolizumab + high-dose cytarabine

II

NCT02768792

R/R AML

 

Nivolumab + cytarabine/

idarubicin

44
(42 with AML)

I/II

NCT02464657

Frontline setting AML, high-risk MDS

CR 64%

(Read the study here)

The combination of immune checkpoint inhibitors with HMAs has demonstrated good activity in preclinical studies and clinical trials are evaluating this combination in patients with AML (Table 3).

Table 3. Clinical trials on immune checkpoint inhibitors in combination with HMAs

AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; ORR, overall response rate; OS, overall survival; R/R, relapsed/refractory

Investigational regimen

N

Phase

Indication

Results

Nivolumab + azacitidine

70

II

R/R AML

ORR 33%

Median OS 6.3 months

(Study covered in the article here)

Ongoing clinical trials

Ipilimumab + decitabine

 

 

I

NCT02890329

R/R AML, R/R MDS

 

Pembrolizumab + decitabine

I/II

NCT02996474

R/R AML

 

Pembrolizumab + azacitidine

II

NCT02845297

R/R AML, R/R MDS, AML ≥ 65 years

 

Nivolumab + azacitidine (arm I)

Nivolumab + azacitidine + ipilimumab (arm II)

II

NCT02397720

R/R AML, AML > 65 years

 

 

Durvalumab + azactidine

213

II

NCT02775903

High-risk MDS, AML ≥ 65 years

(see below)

At the 61st American Society of Hematology (ASH) meeting, the AML Hub spoke to Amer Zeidan, who discussed the results of an ongoing, randomized, study (NCT02775903) in patients (N = 213) with AML or high-risk MDS treated with AZA or AZA plus durvalumab, a PDL-1 inhibitor (video below). Even though the results showed no improvement in ORR, the study provided information on how immune checkpoint blockade plays a role in AML

VIDEO INTERVIEW: What is the role of immune checkpoint directed therapy in the treatment of AML?

What is the role of immune checkpoint directed therapy in the treatment of AML?

New specific immune checkpoint molecules, in addition to CTLA-4, PD-1, and PD-L1, have been identified. In an interview with the AML Hub, Naval Daver talked about unique AML and MDS checkpoint molecules, such as TIM-3, and ongoing clinical trials (video below). Several ongoing clinical trials are studying the combination of different immune checkpoint inhibitors and are reported in Table 4.

Table 4. Ongoing clinical trials on immunotherapy combinations

AML, acute myeloid leukemia; allo-SCT, allogeneic SCT; CR, complete response; MDS myelodysplastic syndrome; R/R relapsed/refractory; SCT, stem cell transplantation

Investigational regimen

Phase

Indication

Nivolumab + ipilimumab

I

NCT01822509

Hematologic malignancies (including AML) relapsed after SCT

Nivolumab (arm I);

ipilimumab (arm II);

nivolumab + ipilimumab (arm III)

I

NCT02846376

AML after allo-SCT

PDR001 (anti-PD-1) + decitabine (arm I);

MBG453 (anti-TIM-3) + decitabine (arm II); PDR001 + MBG453 + decitabine (arm III);

MBG453 (arm IV);

PDR001 + MBG453 (arm V)

I

NCT03066648

R/R AML, R/R MDS, AML ineligible for chemotherapy

Pidilizumab (anti-PD-1) + dendritic cell vaccine

II

NCT01096602

AML in CR

Emerging immunotherapy modalities in AML and exciting data from ASH 2019

Conclusion4

Whilst the use of immune checkpoint inhibitors as monotherapy in AML seems ineffective, their combination with other treatments, such as chemotherapy or HMAs, is promising. Early results from studies evaluating these combinations in both the upfront setting and the R/R AML setting have demonstrated that the addition of immune checkpoint inhibitors to chemotherapy or HMAs is feasible and well-tolerated.

  1. Döhner H, Weisdorf DJ, Bloomfield CD, et al. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136-52. DOI: 1056/NEJMra1406184
  2. Lai C, Doucette K, Norsworthy K. Recent drug approvals for acute myeloid leukemia. J Hematol Oncol. 2019;12:100. DOI: 1186/s13045-019-0774-x
  3. Lichtenegger FS, Krupka C, Haubner S, et al. Recent developments in immunotherapy of acute myeloid leukemia. J Hematol Oncol. 2017;10:142. DOI: 1186/s13045-017-0505-0
  4. Stahl M & Goldberg AD. Immune checkpoint inhibitors in acute myeloid leukemia: Novel combinations and therapeutic targets. Curr Oncol Rep. 2019;21(4):37. DOI: 1007/s11912-019-0781-7

Your opinion matters

Do you intend to implement next-generation sequencing for measurable residual disease monitoring in MDS patients?
1 vote - 2 days left ...

Newsletter

Subscribe to get the best content related to AML delivered to your inbox